1
|
Kolagkis PX, Serviou SK, Stini NA, Demertzidou VP, Poursaitidis ET, Galathri EM, Mountanea OG, Skolia E, Kokotos CG. Deciphering the Knoevenagel condensation: towards a catalyst-free and water-mediated process. Org Biomol Chem 2024; 22:8293-8299. [PMID: 39302224 DOI: 10.1039/d4ob01420k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/22/2024]
Abstract
The Knoevenagel condensation constitutes one of the most well-studied and crucial transformations in organic chemistry, since it facilitates the synthesis of numerous valuable compounds. With the advent of green chemistry, several alternative protocols for the Knoevenagel reaction have been introduced and catalyst-free approaches to the Knoevenagel condensation have also been mentioned, however the harsh temperatures employed and the limited substrate scope restricted their application. Herein, we have performed an extensive study on the catalyst-free and water-mediated Knoevenagel reaction, with specific focus on optimising the green parameters and metrics of our methodology. Additionally, we directly compared our approach with previous catalyst-free methods, while providing a fast assembly of multiple compounds in parallel.
Collapse
Affiliation(s)
- Periklis X Kolagkis
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| | - Stamatis K Serviou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| | - Naya A Stini
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| | - Vera P Demertzidou
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| | - Efthymios T Poursaitidis
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| | - Eirini M Galathri
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| | - Olga G Mountanea
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| | - Elpida Skolia
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| | - Christoforos G Kokotos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, Panepistimiopolis, Athens 15771, Greece.
| |
Collapse
|
2
|
Molitor M, Menge A, Mandel S, George S, Müller S, Knapp S, Hofmann B, Steinhilber D, Häfner AK. Unlocking the potential: unveiling tyrphostins with Michael-reactive cyanoacrylate motif as promising inhibitors of human 5-lipoxygenase. Pflugers Arch 2024:10.1007/s00424-024-03019-7. [PMID: 39347835 DOI: 10.1007/s00424-024-03019-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/01/2024]
Abstract
Human 5-lipoxygenase (5-LO) is the key enzyme in the biosynthesis of leukotrienes, mediators of the innate immune system that also play an important role in inflammatory diseases and cancer. In this study, we present compounds, containing a Michael-reactive cyanoacrylate moiety as potent inhibitors of 5-LO. Representatives of the tyrosine kinase inhibitor family called tyrphostins, structurally related to known 5-LO inhibitors, were screened for their 5-LO inhibitory properties using recombinant human 5-LO, intact human PMNL (polymorphonuclear leukocytes), and PMNL homogenates. Their mode of action was characterized by the addition of glutathione, using a fourfold cysteine 5-LO mutant and mass spectrometry analysis. SAR studies revealed several members of the tyrphostin family containing a Michael-reactive cyanoacrylate to efficiently inhibit 5-LO. We identified degrasyn (IC50 0.11 µM), tyrphostin A9 (IC50 0.8 µM), AG879 (IC50 78 nM), and AG556 (IC50 64 nM) as potent 5-LO inhibitors. Mass spectrometry analysis revealed that degrasyn and AG556 covalently bound to up to four cysteines, including C416 and/or C418 which surround the substrate entry site. Furthermore, the 5-LO inhibitory effect of degrasyn was remarkably impaired by the addition of glutathione or by the mutation of cysteines to serines at the surface of 5-LO. We successfully identified several tyrphostins as potent inhibitors of human 5-LO. Degrasyn and AG556 were able to covalently bind to 5-LO via their cyanoacrylate moiety. This provides a promising mechanism for targeting 5-LO by Michael acceptors, leading to new therapeutic opportunities in the field of inflammation and cancer.
Collapse
Affiliation(s)
- Maximilian Molitor
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany
| | - Amelie Menge
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Max-von-Laue-Str. 15, 60438, Frankfurt Am Main, Germany
| | - Sebastian Mandel
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany
| | - Sven George
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany
| | - Susanne Müller
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Max-von-Laue-Str. 15, 60438, Frankfurt Am Main, Germany
| | - Stefan Knapp
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany
- Buchmann Institute for Molecular Life Sciences and Structural Genomics Consortium (SGC), Max-von-Laue-Str. 15, 60438, Frankfurt Am Main, Germany
| | - Bettina Hofmann
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany
| | - Dieter Steinhilber
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany
| | - Ann-Kathrin Häfner
- Institute of Pharmaceutical Chemistry, Goethe University, Max-von-Laue-Str. 9, 60438, Frankfurt Am Main, Germany.
| |
Collapse
|
3
|
Bhatnagar A, Pemawat G. Anticancer and Antibacterial Activeness of Fused Pyrimidines: Newfangled Updates. Bioorg Chem 2024; 153:107780. [PMID: 39260159 DOI: 10.1016/j.bioorg.2024.107780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 08/14/2024] [Accepted: 08/29/2024] [Indexed: 09/13/2024]
Abstract
Pyrimidine-based heterocyclic compounds are garnering substantial interest due to their essential role as a class of natural and synthetic molecules. These compounds show a diverse array of biologically relevant activities, making them highly prospective candidates for clinical translation as therapeutic agents in combating various diseases. Pyrimidine derivatives and their fused analogues, such as thienopyrimidines, pyrazolopyrimidines, pyridopyrimidines, and pyrimidopyrimidines, hold immense possibility in both anticancer and antibacterial research. These compounds exhibit notable efficacy by targeting protein kinases, which are crucial enzymes regulating fundamental cellular processes like metabolism, migration, division, and growth. Through enzyme inhibition, these derivatives disrupt key cellular signaling pathways, thereby affecting critical cellular functions and viability. The advantage lies in the ubiquity of the pyrimidine structure across various natural compounds, enabling interactions with enzymes, genetic material, and cellular components pivotal for chemical and biological processes. This interaction plays a central role in modulating vital biological activities, making pyrimidine-containing compounds indispensable in drug discovery. In the realm of anticancer therapy, these compounds strategically target key proteins like EGFR, important for aberrant cell growth. Fused pyrimidine motifs, exemplified by various drugs, are designed to inhibit EGFR, thereby impeding tumor progression. Moreover, these compounds influence potent antibacterial activity, interfering with microbial growth through mechanisms ranging from DNA replication inhibition to other vital cellular functions. This dual activity, targeting both cancer cells and microbial pathogens, underscores the versatility and potential of pyrimidine derivatives in medical applications. This review provides insights into the structural characteristics, synthesis methods, and significant medicinal applications of fused pyrimidine derivatives, highlighting their double role in combating cancer and bacterial infections.
Collapse
Affiliation(s)
- Ayushi Bhatnagar
- Department of Chemistry, University College of Science, Mohanlal Sukhadia University, Udaipur, Rajasthan, India 313001
| | - Gangotri Pemawat
- Department of Chemistry, University College of Science, Mohanlal Sukhadia University, Udaipur, Rajasthan, India 313001.
| |
Collapse
|
4
|
Logghe T, van Zwol E, Immordino B, Van den Cruys K, Peeters M, Giovannetti E, Bogers J. Hyperthermia in Combination with Emerging Targeted and Immunotherapies as a New Approach in Cancer Treatment. Cancers (Basel) 2024; 16:505. [PMID: 38339258 PMCID: PMC10854776 DOI: 10.3390/cancers16030505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 01/10/2024] [Accepted: 01/19/2024] [Indexed: 02/12/2024] Open
Abstract
Despite significant advancements in the development of novel therapies, cancer continues to stand as a prominent global cause of death. In many cases, the cornerstone of standard-of-care therapy consists of chemotherapy (CT), radiotherapy (RT), or a combination of both. Notably, hyperthermia (HT), which has been in clinical use in the last four decades, has proven to enhance the effectiveness of CT and RT, owing to its recognized potency as a sensitizer. Furthermore, HT exerts effects on all steps of the cancer-immunity cycle and exerts a significant impact on key oncogenic pathways. Most recently, there has been a noticeable expansion of cancer research related to treatment options involving immunotherapy (IT) and targeted therapy (TT), a trend also visible in the research and development pipelines of pharmaceutical companies. However, the potential results arising from the combination of these innovative therapeutic approaches with HT remain largely unexplored. Therefore, this review aims to explore the oncology pipelines of major pharmaceutical companies, with the primary objective of identifying the principal targets of forthcoming therapies that have the potential to be advantageous for patients by specifically targeting molecular pathways involved in HT. The ultimate goal of this review is to pave the way for future research initiatives and clinical trials that harness the synergy between emerging IT and TT medications when used in conjunction with HT.
Collapse
Affiliation(s)
- Tine Logghe
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Eke van Zwol
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
| | - Benoît Immordino
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, 56127 Pisa, Italy
| | | | - Marc Peeters
- Department of Oncology, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Elisa Giovannetti
- Cancer Pharmacology Lab, Fondazione Pisana per la Scienza, San Giuliano, 56017 Pisa, Italy
- Department of Medical Oncology, Amsterdam UMC, Location Vrije Universiteit, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Johannes Bogers
- Elmedix NV, Dellingstraat 34/1, 2800 Mechelen, Belgium
- Laboratory of Cell Biology and Histology, Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium
| |
Collapse
|
5
|
Mishra A, Yadav P, Awasthi SK. Nitrogen-Enriched Biguanidine-Functionalized Cobalt Ferrite Nanoparticles as a Heterogeneous Base Catalyst for Knoevenagel Condensation under Solvent-Free Conditions. ACS ORGANIC & INORGANIC AU 2023; 3:254-265. [PMID: 37810412 PMCID: PMC10557060 DOI: 10.1021/acsorginorgau.3c00002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 10/10/2023]
Abstract
Designing efficient, economical heterogeneous catalysts for the Knoevenagel condensation reaction is highly significant owing to the importance of reaction products in industries as well as pharmaceutics. Herein, we have designed and synthesized biguanidine-functionalized basic magnetically retrievable cobalt ferrite nanoparticles (CFNPs) for the synthesis of Knoevenagel condensation products using benzaldehydes and active methylene compounds (malononitrile/ethyl cyanoacetate/cyanoacetamide). Several advanced techniques, such as Fourier transform infrared (FT-IR), thermogravimetric analysis (TGA), powder X-ray diffraction (XRD), scanning electron microscopy (SEM), transmission electron microscopy (TEM), and vibration sample magnetometry (VSM), were utilized to precisely characterize the catalyst. The robust features of the current approach involve outstanding catalytic performance, solvent-free reaction conditions, ease of catalyst retrievability, easy workup procedure, large substrate tolerance, high turnover frequency (TOF) values (up to 486.88 h-1), values of green chemistry metrics such as E-factor (0.15), reaction mass efficiency (RME) value (87.07%), carbon efficiency (93.4%), and atom economy (AE) value (88.10%) close to their ideal values, and recyclability up to eight runs without a considerable reduction in activity, boosting the appeal of this approach from a commercial and ecological point of view.
Collapse
Affiliation(s)
| | | | - Satish K. Awasthi
- Chemical Biology Laboratory,
Department of Chemistry, University of Delhi, Delhi 110007, India
| |
Collapse
|
6
|
Yang X, Chen C, Wang K, Chen M, Wang Y, Chen Z, Zhao W, Ou S. Elucidating the molecular mechanisms of ozone therapy for neuropathic pain management by integrated transcriptomic and metabolomic approach. Front Genet 2023; 14:1231682. [PMID: 37779912 PMCID: PMC10536237 DOI: 10.3389/fgene.2023.1231682] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/18/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction: Neuropathic pain remains a prevalent and challenging condition to treat, with current therapies often providing inadequate relief. Ozone therapy has emerged as a promising treatment option; however, its mechanisms of action in neuropathic pain remain poorly understood. Methods: In this study, we investigated the effects of ozone treatment on gene expression and metabolite levels in the brainstem and hypothalamus of a rat model, using a combined transcriptomic and metabolomic approach. Results: Our findings revealed significant alterations in key genes, including DCST1 and AIF1L, and metabolites such as Aconitic acid, L-Glutamic acid, UDP-glucose, and Tyrosine. These changes suggest a complex interplay of molecular pathways and region-specific mechanisms underlying the analgesic effects of ozone therapy. Discussion: Our study provides insights into the molecular targets of ozone treatment for neuropathic pain, laying the groundwork for future research on validating these targets and developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Xiaolan Yang
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chaoming Chen
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Keyang Wang
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Min Chen
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yong Wang
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhengping Chen
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wang Zhao
- Department of Neurology, Yongchuan Hospital Affiliated to Chongqing Medical University, Chongqing, China
| | - Shu Ou
- Department of Neurology, The Fengjie People’s Hospital, Fengjie Branch of the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Pergu R, Shoba VM, Chaudhary SK, Munkanatta Godage DNP, Deb A, Singha S, Dhawa U, Singh P, Anokhina V, Singh S, Siriwardena SU, Choudhary A. Development and Applications of Chimera Platforms for Tyrosine Phosphorylation. ACS CENTRAL SCIENCE 2023; 9:1558-1566. [PMID: 37637727 PMCID: PMC10450875 DOI: 10.1021/acscentsci.3c00200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Indexed: 08/29/2023]
Abstract
Chimeric small molecules that induce post-translational modification (PTM) on a target protein by bringing it into proximity to a PTM-inducing enzyme are furnishing novel modalities to perturb protein function. Despite recent advances, such molecules are unavailable for a critical PTM, tyrosine phosphorylation. Furthermore, the contemporary design paradigm of chimeric molecules, formed by joining a noninhibitory binder of the PTM-inducing enzyme with the binder of the target protein, prohibits the recruitment of most PTM-inducing enzymes as their noninhibitory binders are unavailable. Here, we report two platforms to generate phosphorylation-inducing chimeric small molecules (PHICS) for tyrosine phosphorylation. We generate PHICS from both noninhibitory binders (scantily available, platform 1) and kinase inhibitors (abundantly available, platform 2) using cysteine-based group transfer chemistry. PHICS triggered phosphorylation on tyrosine residues in diverse sequence contexts and target proteins (e.g., membrane-associated, cytosolic) and displayed multiple bioactivities, including the initiation of a growth receptor signaling cascade and the death of drug-resistant cancer cells. These studies provide an approach to induce biologically relevant PTM and lay the foundation for pharmacologic PTM editing (i.e., induction or removal) of target proteins using abundantly available inhibitors of PTM-inducing or -erasing enzymes.
Collapse
Affiliation(s)
- Rajaiah Pergu
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Veronika M. Shoba
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Santosh K. Chaudhary
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | | | - Arghya Deb
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Santanu Singha
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Uttam Dhawa
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Prashant Singh
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Viktoriya Anokhina
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Sameek Singh
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Sachini U. Siriwardena
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Amit Choudhary
- Chemical
Biology and Therapeutics Science, Broad
Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Department
of Medicine, Harvard Medical School, Boston, Massachusetts 02115, United States
- Divisions
of Renal Medicine and Engineering, Brigham
and Women’s Hospital, Boston, Massachusetts 02115, United States
| |
Collapse
|
8
|
Uddin KM, Sakib M, Siraji S, Uddin R, Rahman S, Alodhayb A, Alibrahim KA, Kumer A, Matin MM, Bhuiyan MMH. Synthesis of New Derivatives of Benzylidinemalononitrile and Ethyl 2-Cyano-3-phenylacrylate: In Silico Anticancer Evaluation. ACS OMEGA 2023; 8:25817-25831. [PMID: 37521603 PMCID: PMC10373203 DOI: 10.1021/acsomega.3c01123] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 06/16/2023] [Indexed: 08/01/2023]
Abstract
In this study, microwave-assisted Knoevenagel condensation was used to produce two novel series of derivatives (1-6) from benzylidenemalononitrile and ethyl 2-cyano-3-phenylacrylate. The synthesized compounds were characterized using Fourier transform infrared (FT-IR) and 1H NMR spectroscopies. The pharmacodynamics, toxicity profiles, and biological activities of the compounds were evaluated through an in silico study using prediction of activity spectra for substances (PASS) and Absorption, Distribution, Metabolism, Excretion, and Toxicity (ADMET) studies. According to the PASS prediction results, compounds 1-6 showed greater antineoplastic potency for breast cancer than other types of cancer. Molecular docking was employed to investigate the binding mode and interaction sites of the derivatives (1-6) with three human cancer targets (HER2, EGFR, and human FPPS), and the protein-ligand interactions of these derivatives were compared to those reference standards Tyrphostin 1 (AG9) and Tyrphostin 23 (A23). Compound 3 showed a stronger effect on two cell lines (HER2 and FPPS) than the reference drugs. A 20 ns molecular dynamics (MD) simulation was also conducted to examine the ligand's behavior at the active binding site of the modeled protein, utilizing the lowest docking energy obtained from the molecular docking study. Enthalpies (ΔH), Gibbs free energies (ΔG), entropies (ΔS), and frontier molecular orbital parameters (highest occupied molecular orbital-lowest unoccupied molecular orbital (HOMO-LUMO) gap, hardness, and softness) were calculated to confirm the thermodynamic stability of all derivatives. The consistent results obtained from the in silico studies suggest that compound 3 has potential as a new anticancer and antiparasitic drug. Further research is required to validate its efficacy.
Collapse
Affiliation(s)
- Kabir M. Uddin
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1217, Bangladesh
| | - Mohiuddin Sakib
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1217, Bangladesh
| | - Siam Siraji
- Department
of Biochemistry and Microbiology, North
South University, Bashundhara, Dhaka 1217, Bangladesh
| | - Riaz Uddin
- Biorganic
and Medicinal Chemistry Laboratory, Department of Chemistry, University of Chittagong, Chattogram 4331, Bangladesh
| | - Shofiur Rahman
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
| | - Abdullah Alodhayb
- Biological
and Environmental Sensing Research Unit, King Abdullah Institute for
Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia
- Research
Chair for Tribology, Surface, and Interface Sciences, Department of
Physics and Astronomy, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Khuloud A. Alibrahim
- Department
of Chemistry, College of Science, Princess
Nourah bint Abdulrahman University, P.O. Box 84428, Riyadh 11671, Saudi Arabia
| | - Ajoy Kumer
- Department
of Chemistry, European University of Bangladesh, Gabtoli, Dhaka 1216, Bangladesh
| | - M. Mahbubul Matin
- Biorganic
and Medicinal Chemistry Laboratory, Department of Chemistry, University of Chittagong, Chattogram 4331, Bangladesh
| | - Md. Mosharef H. Bhuiyan
- Biorganic
and Medicinal Chemistry Laboratory, Department of Chemistry, University of Chittagong, Chattogram 4331, Bangladesh
| |
Collapse
|
9
|
Heininen J, Erbacher C, Kotiaho T, Kostiainen R, Teppo J. Enzymatic Phosphorylation of Oxidized Tyrosine Residues. J Proteome Res 2023. [PMID: 37146082 DOI: 10.1021/acs.jproteome.3c00061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Post-translational modifications (PTMs) alter the function and fate of proteins and cells in almost every conceivable way. Protein modifications can occur as a result of specific regulating actions of enzymes, such as tyrosine kinases phosphorylating tyrosine residues or by nonenzymatic reactions, such as oxidation related to oxidative stress and diseases. While many studies have addressed the multisite, dynamic, and network-like properties of PTMs, only little is known of the interplay of the same site modifications. In this work, we studied the enzymatic phosphorylation of oxidized tyrosine (l-DOPA) residues using synthetic insulin receptor peptides, in which tyrosine residues were replaced with l-DOPA. The phosphorylated peptides were identified by liquid chromatography-high-resolution mass spectrometry and the site of phosphorylation by tandem mass spectrometry. The results clearly show that the oxidized tyrosine residues are phosphorylated, displaying a specific immonium ion peak in the MS2 spectra. Furthermore, we detected this modification in our reanalysis (MassIVE ID: MSV000090106) of published bottom-up phosphoproteomics data. The modification, where both oxidation and phosphorylation take place at the same amino acid, has not yet been published in PTM databases. Our data indicate that there can be multiple PTMs that do not exclude each other at the same modification site.
Collapse
Affiliation(s)
- Juho Heininen
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
| | - Catharina Erbacher
- Institute of Inorganic and Analytical Chemistry, University of Münster, Corrensstraße 48, 48149 Münster, Germany
| | - Tapio Kotiaho
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
- Department of Chemistry, Faculty of Science, University of Helsinki, P.O. Box 55, FI-00014 Helsinki, Finland
| | - Risto Kostiainen
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
| | - Jaakko Teppo
- Drug Research Program and Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, P.O. Box 56, FI-00014 Helsinki, Finland
| |
Collapse
|
10
|
Yadav TT, Moin Shaikh G, Kumar MS, Chintamaneni M, YC M. A Review on Fused Pyrimidine Systems as EGFR Inhibitors and Their Structure–Activity Relationship. Front Chem 2022; 10:861288. [PMID: 35769445 PMCID: PMC9234326 DOI: 10.3389/fchem.2022.861288] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 04/28/2022] [Indexed: 01/05/2023] Open
Abstract
Epidermal growth factor receptor (EGFR) belongs to the family of tyrosine kinase that is activated when a specific ligand binds to it. The EGFR plays a vital role in the cellular proliferation process, differentiation, and apoptosis. In the case of cancer, EGFR undergoes uncontrolled auto-phosphorylation that results in increased cellular proliferation and decreased apoptosis, causing cancer promotion. From the literature, it shows that pyrimidine is one of the most commonly studied heterocycles for its antiproliferative activity against EGFR inhibition. The authors have collated some interesting results in the heterocycle-fused pyrimidines that have been studied using different cell lines (sensitive and mutational) and in animal models to determine their activity and potency. It is quite clear that the fused systems are highly effective in inhibiting EGFR activity in cancer cells. Therefore, the structure–activity relationship (SAR) comes into play in determining the nature of the heterocycle and the substituents that are responsible for the increased activity and toxicity. Understanding the SAR of heterocycle-fused pyrimidines will help in getting a better overview of the molecules concerning their activity and potency profile as future EGFR inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Mayur YC
- *Correspondence: Mayur YC, mayur
| |
Collapse
|
11
|
Malekan M, Ebrahimzadeh MA. Vascular Endothelial Growth Factor Receptors [VEGFR] as Target in Breast Cancer Treatment: Current Status in Preclinical and Clinical Studies and Future Directions. Curr Top Med Chem 2022; 22:891-920. [PMID: 35260067 DOI: 10.2174/1568026622666220308161710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/11/2022] [Accepted: 01/20/2022] [Indexed: 12/09/2022]
Abstract
Breast cancer [BC] is one of the most common cancers among women, one of the leading causes of a considerable number of cancer-related death globally. Among all procedures leading to the formation of breast tumors, angiogenesis has an important role in cancer progression and outcomes. Therefore, various anti-angiogenic strategies have developed so far to enhance treatment's efficacy in different types of BC. Vascular endothelial growth factors [VEGFs] and their receptors are regarded as the most well-known regulators of neovascularization. VEGF binding to vascular endothelial growth factor receptors [VEGFRs] provides cell proliferation and vascular tissue formation by the subsequent tyrosine kinase pathway. VEGF/VEGFR axis displays an attractive target for anti-angiogenesis and anti-cancer drug design. This review aims to describe the existing literature regarding VEGFR inhibitors, focusing on BC treatment reported in the last two decades.
Collapse
Affiliation(s)
- Mohammad Malekan
- Student Research Committee, School of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Ali Ebrahimzadeh
- Pharmaceutical Sciences Research Center, School of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
12
|
Pereira Moreira B, Weber MHW, Haeberlein S, Mokosch AS, Spengler B, Grevelding CG, Falcone FH. Drug Repurposing and De Novo Drug Discovery of Protein Kinase Inhibitors as New Drugs against Schistosomiasis. Molecules 2022; 27:molecules27041414. [PMID: 35209202 PMCID: PMC8879451 DOI: 10.3390/molecules27041414] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 02/09/2022] [Accepted: 02/16/2022] [Indexed: 02/01/2023] Open
Abstract
Schistosomiasis is a neglected tropical disease affecting more than 200 million people worldwide. Chemotherapy relies on one single drug, praziquantel, which is safe but ineffective at killing larval stages of this parasite. Furthermore, concerns have been expressed about the rise in resistance against this drug. In the absence of an antischistosomal vaccine, it is, therefore, necessary to develop new drugs against the different species of schistosomes. Protein kinases are important molecules involved in key cellular processes such as signaling, growth, and differentiation. The kinome of schistosomes has been studied and the suitability of schistosomal protein kinases as targets demonstrated by RNA interference studies. Although protein kinase inhibitors are mostly used in cancer therapy, e.g., for the treatment of chronic myeloid leukemia or melanoma, they are now being increasingly explored for the treatment of non-oncological conditions, including schistosomiasis. Here, we discuss the various approaches including screening of natural and synthetic compounds, de novo drug development, and drug repurposing in the context of the search for protein kinase inhibitors against schistosomiasis. We discuss the status quo of the development of kinase inhibitors against schistosomal serine/threonine kinases such as polo-like kinases (PLKs) and mitogen-activated protein kinases (MAP kinases), as well as protein tyrosine kinases (PTKs).
Collapse
Affiliation(s)
- Bernardo Pereira Moreira
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
| | - Michael H. W. Weber
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
| | - Simone Haeberlein
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
| | - Annika S. Mokosch
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (A.S.M.); (B.S.)
| | - Bernhard Spengler
- Institute of Inorganic and Analytical Chemistry, Justus Liebig University Giessen, 35392 Giessen, Germany; (A.S.M.); (B.S.)
| | - Christoph G. Grevelding
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
| | - Franco H. Falcone
- Biomedical Research Center Seltersberg (BFS), Institute of Parasitology, Justus Liebig University Giessen, 35392 Giessen, Germany; (B.P.M.); (M.H.W.W.); (S.H.); (C.G.G.)
- Correspondence:
| |
Collapse
|
13
|
Phosphorylation of TRIP13 at Y56 induces radiation resistance but sensitizes head and neck cancer to cetuximab. Mol Ther 2022; 30:468-484. [PMID: 34111559 PMCID: PMC8753291 DOI: 10.1016/j.ymthe.2021.06.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 03/10/2021] [Accepted: 06/01/2021] [Indexed: 01/07/2023] Open
Abstract
Radiation therapy, a mainstay of treatment for head and neck cancer, is not always curative due to the development of treatment resistance; additionally, multi-institutional trials have questioned the efficacy of concurrent radiation with cetuximab, the epidermal growth factor receptor (EGFR) inhibitor. We unraveled a mechanism for radiation resistance; that is, radiation induces EGFR, which phosphorylates TRIP13 (thyroid hormone receptor interactor 13) on tyrosine 56. Phosphorylated (phospho-)TRIP13 promotes non-homologous end joining (NHEJ) repair to induce radiation resistance. NHEJ is the main repair pathway for radiation-induced DNA damage. Tumors expressing high TRIP13 do not respond to radiation but are sensitive to cetuximab or cetuximab combined with radiation. Suppression of phosphorylation of TRIP13 at Y56 abrogates these effects. These findings show that EGFR-mediated phosphorylation of TRIP13 at Y56 is a vital mechanism of radiation resistance. Notably, TRIP13-pY56 could be used to predict the response to radiation or cetuximab and could be explored as an actionable target.
Collapse
|
14
|
Gutiérrez S, Tomás‐Gamasa M, Mascareñas JL. Exporting Metal‐Carbene Chemistry to Live Mammalian Cells: Copper‐Catalyzed Intracellular Synthesis of Quinoxalines Enabled by N−H Carbene Insertions. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202108899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Sara Gutiérrez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS) Departamento de Química Orgánica Universidade de Santiago de Compostela 15705 Santiago de Compostela Spain
| | - María Tomás‐Gamasa
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS) Departamento de Química Orgánica Universidade de Santiago de Compostela 15705 Santiago de Compostela Spain
| | - José L. Mascareñas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS) Departamento de Química Orgánica Universidade de Santiago de Compostela 15705 Santiago de Compostela Spain
| |
Collapse
|
15
|
Gutiérrez S, Tomás‐Gamasa M, Mascareñas JL. Exporting Metal-Carbene Chemistry to Live Mammalian Cells: Copper-Catalyzed Intracellular Synthesis of Quinoxalines Enabled by N-H Carbene Insertions. Angew Chem Int Ed Engl 2021; 60:22017-22025. [PMID: 34390304 PMCID: PMC8518842 DOI: 10.1002/anie.202108899] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Indexed: 12/17/2022]
Abstract
Implementing catalytic organometallic transformations in living settings can offer unprecedented opportunities in chemical biology and medicine. Unfortunately, the number of biocompatible reactions so far discovered is very limited, and essentially restricted to uncaging processes. Here, we demonstrate the viability of performing metal carbene transfer reactions in live mammalian cells. In particular, we show that copper (II) catalysts can promote the intracellular annulation of alpha-keto diazocarbenes with ortho-amino arylamines, in a process that is initiated by an N-H carbene insertion. The potential of this transformation is underscored by the in cellulo synthesis of a product that alters mitochondrial functions, and by demonstrating cell selective biological responses using targeted copper catalysts. Considering the wide reactivity spectrum of metal carbenes, this work opens the door to significantly expanding the repertoire of life-compatible abiotic reactions.
Collapse
Affiliation(s)
- Sara Gutiérrez
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS)Departamento de Química OrgánicaUniversidade de Santiago de Compostela15705Santiagode CompostelaSpain
| | - María Tomás‐Gamasa
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS)Departamento de Química OrgánicaUniversidade de Santiago de Compostela15705Santiagode CompostelaSpain
| | - José L. Mascareñas
- Centro Singular de Investigación en Química Biolóxica e Materiais Moleculares (CiQUS)Departamento de Química OrgánicaUniversidade de Santiago de Compostela15705Santiagode CompostelaSpain
| |
Collapse
|
16
|
Wilson BAP, Voeller D, Smith EA, Wamiru A, Goncharova EI, Liu G, Lipkowitz S, O’Keefe BR. In Vitro Ubiquitination Platform Identifies Methyl Ellipticiniums as Ubiquitin Ligase Inhibitors. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2021; 26:870-884. [PMID: 33882749 PMCID: PMC9907454 DOI: 10.1177/24725552211000675] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The transfer of the small protein ubiquitin to a target protein is an intricately orchestrated process called ubiquitination that results in modulation of protein function or stability. Proper regulation of ubiquitination is essential, and dysregulation of this process is implicated in several human diseases. An example of a ubiquitination cascade that is a central signaling node in important disease-associated pathways is that of CBLB [a human homolog of a viral oncogene Casitas B-lineage lymphoma (CBL) from the Cas NS-1 murine retrovirus], a RING finger ubiquitin ligase (E3) whose substrates include a number of important cell-signaling kinases. These include kinases important in immune function that act in the T cell receptor and costimulatory pathways, the Tyro/Axl/MerTK (TAM) receptor family in natural killer (NK) cells, as well as growth factor receptor kinases like epidermal growth factor receptor (EGFR). Loss of CBLB has been shown to increase innate and adaptive antitumor immunity. This suggests that small-molecule modulation of CBLB E3 activity could enhance antitumor immunity in patients. To explore the hypothesis that enzymatic inhibition of E3s may result in modulation of disease-related signaling pathways, we established a high-throughput screen of >70,000 chemical entities for inhibition of CBLB activity. Although CBLB was chosen as a proof-of-principle target for inhibitor discovery, we demonstrate that our assay is generalizable to monitoring the activity of other ubiquitin ligases. We further extended our observed in vitro inhibition with additional cell-based models of CBLB activity. From these studies, we demonstrate that a class of natural product-based alkaloids, known as methyl ellipticiniums (MEs), is capable of inhibiting ubiquitin ligases intracellularly.
Collapse
Affiliation(s)
- Brice A. P. Wilson
- Molecular Targets Program, Center for Cancer Research,
National Cancer Institute, Frederick, MD, USA
| | - Donna Voeller
- Women’s Malignancies Branch, Center for Cancer
Research, National Cancer Institute, National Institutes of Health, Bethesda, MD,
USA
| | - Emily A. Smith
- Molecular Targets Program, Center for Cancer Research,
National Cancer Institute, Frederick, MD, USA
- Basic Science Program, Leidos Biomedical Research,
Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Antony Wamiru
- Molecular Targets Program, Center for Cancer Research,
National Cancer Institute, Frederick, MD, USA
- Basic Science Program, Leidos Biomedical Research,
Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Ekaterina I. Goncharova
- Molecular Targets Program, Center for Cancer Research,
National Cancer Institute, Frederick, MD, USA
- Advanced Biomedical Computational Science, Frederick
National Laboratory for Cancer Research, Frederick, MD, USA
| | - Gang Liu
- Department of Pharmacology and Pharmaceutical Sciences,
School of Medicine, Institute of Materia Medica, Chinese Academy of Medical
Sciences, Peking Union Medical College, Tsinghua-Peking Center for Life Sciences,
Tsinghua University, Beijing, China
| | - Stanley Lipkowitz
- Women’s Malignancies Branch, Center for Cancer
Research, National Cancer Institute, National Institutes of Health, Bethesda, MD,
USA
| | - Barry R. O’Keefe
- Molecular Targets Program, Center for Cancer Research,
National Cancer Institute, Frederick, MD, USA
- Natural Products Branch, Developmental Therapeutics
Program, Division of Cancer Treatment and Diagnosis, National Cancer Institute,
Frederick, MD, USA
| |
Collapse
|
17
|
Pacini L, Jenks AD, Lima NC, Huang PH. Targeting the Fibroblast Growth Factor Receptor (FGFR) Family in Lung Cancer. Cells 2021; 10:1154. [PMID: 34068816 PMCID: PMC8151052 DOI: 10.3390/cells10051154] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 12/12/2022] Open
Abstract
Lung cancer is the most common cause of cancer-related deaths globally. Genetic alterations, such as amplifications, mutations and translocations in the fibroblast growth factor receptor (FGFR) family have been found in non-small cell lung cancer (NSCLC) where they have a role in cancer initiation and progression. FGFR aberrations have also been identified as key compensatory bypass mechanisms of resistance to targeted therapy against mutant epidermal growth factor receptor (EGFR) and mutant Kirsten rat sarcoma 2 viral oncogene homolog (KRAS) in lung cancer. Targeting FGFR is, therefore, of clinical relevance for this cancer type, and several selective and nonselective FGFR inhibitors have been developed in recent years. Despite promising preclinical data, clinical trials have largely shown low efficacy of these agents in lung cancer patients with FGFR alterations. Preclinical studies have highlighted the emergence of multiple intrinsic and acquired resistance mechanisms to FGFR tyrosine kinase inhibitors, which include on-target FGFR gatekeeper mutations and activation of bypass signalling pathways and alternative receptor tyrosine kinases. Here, we review the landscape of FGFR aberrations in lung cancer and the array of targeted therapies under clinical evaluation. We also discuss the current understanding of the mechanisms of resistance to FGFR-targeting compounds and therapeutic strategies to circumvent resistance. Finally, we highlight our perspectives on the development of new biomarkers for stratification and prediction of FGFR inhibitor response to enable personalisation of treatment in patients with lung cancer.
Collapse
Affiliation(s)
| | | | | | - Paul H. Huang
- Division of Molecular Pathology, The Institute of Cancer Research, London SM2 5NG, UK; (L.P.); (A.D.J.); (N.C.L.)
| |
Collapse
|
18
|
Zucchiatti P, Birarda G, Cerea A, Semrau MS, Hubarevich A, Storici P, De Angelis F, Toma A, Vaccari L. Binding of tyrosine kinase inhibitor to epidermal growth factor receptor: surface-enhanced infrared absorption microscopy reveals subtle protein secondary structure variations. NANOSCALE 2021; 13:7667-7677. [PMID: 33928964 DOI: 10.1039/d0nr09200b] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Surface-Enhanced Infrared Absorption (SEIRA) has been proposed as a valuable tool for protein binding studies, but its performances have been often proven on model proteins undergoing severe secondary structure rearrangements, while ligand binding only marginally involves the protein backbone in the vast majority of the biologically relevant cases. In this study we demonstrate the potential of SEIRA microscopy for highlighting the very subtle secondary structure modifications associated with the binding of Lapatinib, a tyrosine kinase inhibitor (TKI), to epidermal growth factor receptor (EGFR), a well-known driver of tumorigenesis in pathological settings such as lung, breast and brain cancers. By boosting the performances of Mid-IR plasmonic devices based on nanoantennas cross-geometry, accustoming the protein purification protocols, carefully tuning the protein anchoring methodology and optimizing the data analysis, we were able to detect EGFR secondary structure modification associated with few amino acids. A nano-patterned platform with this kind of sensitivity bridges biophysical and structural characterization methods, thus opening new possibilities in studying of proteins of biomedical interest, particularly for drug-screening purposes.
Collapse
Affiliation(s)
- Paolo Zucchiatti
- Elettra Sincrotrone Trieste SCpA, S.S. 14 Km 163.5, I-34149, Basovizza, Trieste, Italy. and Universtà degli studi di Trieste, Dipartimento di Fisica, via Valerio 2, I-34127, Trieste, Italy
| | - Giovanni Birarda
- Elettra Sincrotrone Trieste SCpA, S.S. 14 Km 163.5, I-34149, Basovizza, Trieste, Italy.
| | - Andrea Cerea
- Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Marta S Semrau
- Elettra Sincrotrone Trieste SCpA, S.S. 14 Km 163.5, I-34149, Basovizza, Trieste, Italy.
| | | | - Paola Storici
- Elettra Sincrotrone Trieste SCpA, S.S. 14 Km 163.5, I-34149, Basovizza, Trieste, Italy.
| | | | - Andrea Toma
- Istituto Italiano di Tecnologia, Via Morego 30, I-16163, Genova, Italy
| | - Lisa Vaccari
- Elettra Sincrotrone Trieste SCpA, S.S. 14 Km 163.5, I-34149, Basovizza, Trieste, Italy.
| |
Collapse
|
19
|
Riccardi C, Napolitano E, Platella C, Musumeci D, Melone MAB, Montesarchio D. Anti-VEGF DNA-based aptamers in cancer therapeutics and diagnostics. Med Res Rev 2020; 41:464-506. [PMID: 33038031 DOI: 10.1002/med.21737] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Revised: 09/12/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022]
Abstract
The vascular endothelial growth factor (VEGF) family and its receptors play fundamental roles not only in physiological but also in pathological angiogenesis, characteristic of cancer progression. Aiming at finding putative treatments for several malignancies, various small molecules, antibodies, or protein-based drugs have been evaluated in vitro and in vivo as VEGF inhibitors, providing efficient agents approved for clinical use. Due to the high clinical importance of VEGF, also a great number of anti-VEGF nucleic acid-based aptamers-that is, oligonucleotides able to bind with high affinity and specificity a selected biological target-have been developed as promising agents in anticancer strategies. Notable research efforts have been made in optimization processes of the identified aptamers, searching for increased target affinity and/or bioactivity by exploring structural analogues of the lead compounds. This review is focused on recent studies devoted to the development of DNA-based aptamers designed to target VEGF. Their therapeutic potential as well as their significance in the construction of highly selective biosensors is here discussed.
Collapse
Affiliation(s)
- Claudia Riccardi
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.,Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Inter-University Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples, Italy
| | - Ettore Napolitano
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Chiara Platella
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Domenica Musumeci
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.,Institute of Biostructures and Bioimages, Naples, Italy
| | - Mariarosa A B Melone
- Department of Advanced Medical and Surgical Sciences, 2nd Division of Neurology, Center for Rare Diseases and Inter-University Center for Research in Neurosciences, University of Campania Luigi Vanvitelli, Naples, Italy.,Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, Pennsylvania, USA
| | - Daniela Montesarchio
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| |
Collapse
|
20
|
Zengin N, Burhan H, Şavk A, Göksu H, Şen F. Synthesis of benzylidenemalononitrile by Knoevenagel condensation through monodisperse carbon nanotube-based NiCu nanohybrids. Sci Rep 2020; 10:12758. [PMID: 32728177 PMCID: PMC7391679 DOI: 10.1038/s41598-020-69764-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 07/20/2020] [Indexed: 11/16/2022] Open
Abstract
Monodisperse nickel/copper nanohybrids (NiCu@MWCNT) based on multi-walled carbon nanotubes (MWCNT) were prepared for the Knoevenagel condensation of aryl and aliphatic aldehydes. The synthesis of these nanohybrids was carried out by the ultrasonic hydroxide assisted reduction method. NiCu@MWCNT nanohybrids were characterized by analytical techniques such as X-ray diffraction (XRD), X-ray photoelectron spectroscopy (XPS), transmission electron microscopy (TEM), high-resolution transmission electron microscopy (HR-TEM), and Raman spectroscopy. According to characterization results, NiCu@MWCNT showed that these nanohybrids form highly uniform, crystalline, monodisperse, colloidally stable NiCu@MWCNT nanohybrids were successfully synthesized. Thereafter, a model reaction was carried out to obtain benzylidenemalononitrile derivatives using NiCu@MWCNT as a catalyst, and showed high catalytic performance under mild conditions over 10-180 min.
Collapse
Affiliation(s)
- Nursefa Zengin
- Kaynasli Vocational College, Duzce University, Düzce, 81900, Turkey
| | - Hakan Burhan
- Sen Research Group, Biochemistry Department, Faculty of Arts and Science, Dumlupınar University, Evliya Çelebi Campus, 43100, Kütahya, Turkey
| | - Aysun Şavk
- Sen Research Group, Biochemistry Department, Faculty of Arts and Science, Dumlupınar University, Evliya Çelebi Campus, 43100, Kütahya, Turkey
| | - Haydar Göksu
- Kaynasli Vocational College, Duzce University, Düzce, 81900, Turkey.
| | - Fatih Şen
- Sen Research Group, Biochemistry Department, Faculty of Arts and Science, Dumlupınar University, Evliya Çelebi Campus, 43100, Kütahya, Turkey.
| |
Collapse
|
21
|
Hiller NDJ, do Amaral e Silva NA, Tavares TA, Faria RX, Eberlin MN, de Luna Martins D. Arylboronic Acids and their Myriad of Applications Beyond Organic Synthesis. European J Org Chem 2020. [DOI: 10.1002/ejoc.202000396] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Noemi de Jesus Hiller
- Instituto de Química; Laboratório de Catálise e Síntese (Lab CSI); Laboratório 413; Universidade Federal Fluminense; Outeiro de São João Batista s/n; Campus do Valonguinho, Centro Niterói RJ 24020-141 Brasil
| | - Nayane Abreu do Amaral e Silva
- Instituto de Química; Laboratório de Catálise e Síntese (Lab CSI); Laboratório 413; Universidade Federal Fluminense; Outeiro de São João Batista s/n; Campus do Valonguinho, Centro Niterói RJ 24020-141 Brasil
| | - Thais Apolinário Tavares
- Instituto de Química; Laboratório de Catálise e Síntese (Lab CSI); Laboratório 413; Universidade Federal Fluminense; Outeiro de São João Batista s/n; Campus do Valonguinho, Centro Niterói RJ 24020-141 Brasil
| | - Robson Xavier Faria
- Laboratório de Toxoplasmose e outras Protozooses; Instituto Oswaldo Cruz, Fiocruz; Av. Brasil, 4365 Manguinhos Rio de Janeiro RJ 21040-360 Brasil
| | - Marcos Nogueira Eberlin
- Mackenzie Presbyterian University; School of Engineering; Rua da Consolação, 930 SP 01302-907 São Paulo Brasil
| | - Daniela de Luna Martins
- Instituto de Química; Laboratório de Catálise e Síntese (Lab CSI); Laboratório 413; Universidade Federal Fluminense; Outeiro de São João Batista s/n; Campus do Valonguinho, Centro Niterói RJ 24020-141 Brasil
| |
Collapse
|
22
|
Characterization of Atypical Protein Tyrosine Kinase (PTK) Genes and Their Role in Abiotic Stress Response in Rice. PLANTS 2020; 9:plants9050664. [PMID: 32456239 PMCID: PMC7284356 DOI: 10.3390/plants9050664] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 02/02/2023]
Abstract
Tyrosine phosphorylation constitutes up to 5% of the total phophoproteome. However, only limited studies are available on protein tyrosine kinases (PTKs) that catalyze protein tyrosine phosphorylation in plants. In this study, domain analysis of the 27 annotated PTK genes in rice genome led to the identification of 18 PTKs with tyrosine kinase domain. The kinase domain of rice PTKs shared high homology with that of dual specificity kinase BRASSINOSTEROID-INSENSITIVE 1 (BRI1) of Arabidopsis. In phylogenetic analysis, rice PTKs clustered with receptor-like cytoplasmic kinases-VII (RLCKs-VII) of Arabidopsis. mRNAseq analysis using Genevestigator revealed that rice PTKs except PTK9 and PTK16 express at moderate to high level in most tissues. PTK16 expression was highly abundant in panicle at flowering stage. mRNAseq data analysis led to the identification of drought, heat, salt, and submergence stress regulated PTK genes in rice. PTK14 was upregulated under all stresses. qRT-PCR analysis also showed that all PTKs except PTK10 were significantly upregulated in root under osmotic stress. Tissue specificity and abiotic stress mediated differential regulation of PTKs suggest their potential role in development and stress response of rice. The candidate dual specificity PTKs identified in this study paves way for molecular analysis of tyrosine phosphorylation in rice.
Collapse
|
23
|
Rutkowska A, Stoczyńska-Fidelus E, Janik K, Włodarczyk A, Rieske P. EGFR vIII: An Oncogene with Ambiguous Role. JOURNAL OF ONCOLOGY 2019; 2019:1092587. [PMID: 32089685 PMCID: PMC7024087 DOI: 10.1155/2019/1092587] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 11/22/2019] [Indexed: 12/21/2022]
Abstract
Epidermal growth factor receptor variant III (EGFRvIII) seems to constitute the perfect therapeutic target for glioblastoma (GB), as it is specifically present on up to 28-30% of GB cells. In case of other tumor types, expression and possible role of this oncogene still remain controversial. In spite of EGFRvIII mechanism of action being crucial for the design of small active anticancer molecules and immunotherapies, i.e., CAR-T technology, it is yet to be precisely defined. EGFRvIII is known to be resistant to degradation, but it is still unclear whether it heterodimerizes with EGF-activated wild-type EGFR (EGFRWT) or homodimerizes (including covalent homodimerization). Constitutive kinase activity of this mutated receptor is relatively low, and some researchers even claim that a nuclear, but not a membrane function, is crucial for its activity. Based on the analyses of recurrent tumors that are often lacking EGFRvIII expression despite its initial presence in corresponding primary foci, this oncogene is suggested to play a marginal role during later stages of carcinogenesis, while even in primary tumors EGFRvIII expression is detected only in a small percentage of tumor cells, undermining the rationality of EGFRvIII-targeting therapies. On the other hand, EGFRvIII-positive cells are resistant to apoptosis, more invasive, and characterized with enhanced proliferation rate. Moreover, expression of this oncogenic receptor was also postulated to be a marker of cancer stem cells. Opinions regarding the role that EGFRvIII plays in tumorigenesis and for tumor aggressiveness are clearly contradictory and, therefore, it is crucial not only to determine its mechanism of action, but also to unambiguously define its role at early and advanced cancer stages.
Collapse
Affiliation(s)
- Adrianna Rutkowska
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Ewelina Stoczyńska-Fidelus
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Research and Development, Celther Polska Ltd., Milionowa 23, 93-193 Lodz, Poland
- Department of Research and Development, Personather Ltd., Milionowa 23, 93-193 Lodz, Poland
| | - Karolina Janik
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Aneta Włodarczyk
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Piotr Rieske
- Department of Tumor Biology, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
- Department of Research and Development, Celther Polska Ltd., Milionowa 23, 93-193 Lodz, Poland
- Department of Research and Development, Personather Ltd., Milionowa 23, 93-193 Lodz, Poland
| |
Collapse
|
24
|
Nanoformulations of small molecule protein tyrosine kinases inhibitors potentiate targeted cancer therapy. Int J Pharm 2019; 573:118785. [PMID: 31678384 DOI: 10.1016/j.ijpharm.2019.118785] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/05/2019] [Accepted: 10/10/2019] [Indexed: 01/08/2023]
Abstract
Protein tyrosine kinases (PTKs) are closely related to tumor development and usually participate in apoptosis, DNA repair, and cell proliferation by activating signaling pathways. Therefore, PTKs have become the most promising targets for cancer therapy. In recent years, a large number of studies on the mechanism of tyrosine kinase activation have indicated that tyrosine kinase inhibitors (TKIs) have important clinical significance and application prospects as targeted anticancer drugs because they can effectively block certain cellular signaling pathways, inhibit tumor metastases and reduce tumor proliferation. Although the increasing emergence of anticancer drug resistance limits the clinical application of TKIs, emerging nanotechnology has made it possible to solve this problem. In this work, the state-of-art of small molecule protein tyrosine kinase inhibitors and the applications of drug delivery systems for TKIs are reviewed, and the potentials and challenges for future research of small molecule TKIs are addressed.
Collapse
|
25
|
Kreisman NR, Wooliscroft LB, Campbell CF, Dotiwala AK, Cox ML, Denson AC, Betancourt AM, Tomchuck SL. Preconditioning hippocampal slices with hypothermia promotes rapid tolerance to hypoxic depolarization and swelling: Mediation by erythropoietin. Brain Res 2019; 1726:146517. [PMID: 31634451 DOI: 10.1016/j.brainres.2019.146517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 08/06/2019] [Accepted: 10/17/2019] [Indexed: 12/26/2022]
Abstract
We suggested previously that hippocampal slices were protected from hypoxic depolarization and swelling by preincubating them at room temperature (Kreisman et al., 2000). We postulated that hypothermic preconditioning induced tolerance in our slices, which protected against hypoxic depolarization and swelling. Control hippocampal slices were incubated at 34-35 °C for two hours and the response to 10 min of severe hypoxia was compared to slices which were preconditioned for two hours at room temperature (22-23 °C) prior to warming to 34-35 °C. Recordings of the extracellular DC potential provided an index of tissue depolarization and changes in tissue light transmittance provided an index of swelling. Hypothermic preconditioning significantly reduced hypoxia-induced swelling, particularly in CA3 and the dentate inner blade. Since erythropoietin (EPO) had been shown to mediate hypoxic preconditioning, we tested whether EPO also mediated hypothermic preconditioning in our slices. Recombinant rat EPO (1-10 micromolar) mitigated hypoxia-induced swelling and depolarization in dentate inner blade of unconditioned slices in a dose-dependent manner. We also blocked the protective effects of hypothermic preconditioning on hypoxic depolarization and swelling in the inner blade of the dentate gyrus by administering soluble EPO receptor in the bath and treating slices with wortmannin to block phosphorylation of PI3 kinase, a critical step in the activation of the downstream neuroprotectant, Akt. These results suggest that EPO mediates tolerance to hypoxic depolarization and swelling induced by hypothermic preconditioning. They also emphasize that various preincubation protocols used in experiments with hippocampal slices may differentially affect basal electrophysiological and metabolic properties of those slices.
Collapse
Affiliation(s)
- Norman R Kreisman
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States; Tulane Brain Institute, New Orleans, LA 70118, United States.
| | | | - Carolyn F Campbell
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States; Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Ary K Dotiwala
- Tulane Brain Institute, New Orleans, LA 70118, United States
| | - Michael L Cox
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Aaron C Denson
- Department of Physiology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Aline M Betancourt
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, United States
| | - Suzanne L Tomchuck
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, United States
| |
Collapse
|
26
|
Kandandapani S, Ridzwan NFW, Mohamad SB, Tayyab S. Exploring the interaction between tyrphostin 9 and human serum albumin using biophysical and computational methods. J Biomol Struct Dyn 2019; 38:4134-4142. [PMID: 31552810 DOI: 10.1080/07391102.2019.1673210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Tyrphostin 9 (Tyr 9) is a potent platelet-derived growth factor receptor (PDGFR) inhibitor, which induces apoptosis in various cancer cell types. The binding of Tyr 9 to the major transport protein, human serum albumin (HSA) was investigated using several spectroscopic techniques and molecular docking method. Fluorescence quenching titration results showed progressive decrease in the protein fluorescence with increasing drug concentrations. A decreasing trend of the Stern-Volmer constant, K sv with increasing temperature characterized the drug-induced quenching as static quenching, thus pointed towards the formation of Tyr 9-HSA complex. The binding constant of Tyr 9-HSA interaction was found to lie within the range 3.48-1.69 × 105 M-1 at three different temperatures, i.e. 15 °C, 25 °C and 35 °C, respectively and suggested intermediate binding affinity between Tyr 9 and HSA. The drug-HSA complex seems to be stabilized by hydrophobic forces, van der Waals forces and hydrogen bonds, as suggested from the thermodynamic data as well as molecular docking results. The far-UV and the near-UV CD spectral results showed slight alteration in the secondary and tertiary structures, respectively, of the protein upon Tyr 9 binding. Interaction of Tyr 9 with HSA also produced microenvironmental perturbations around protein fluorophores, as evident from the three-dimensional fluorescence spectral results but increased protein's thermal stability. Both competitive drug binding results and molecular docking analysis suggested Sudlow's Site I of HSA as the preferred Tyr 9 binding site. Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Salanee Kandandapani
- Biochemistry Programme, Biomolecular Research Group, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Nor Farrah Wahidah Ridzwan
- Bioinformatics Programme, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia
| | - Saharuddin B Mohamad
- Bioinformatics Programme, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia.,Centre of Research for Computational Sciences and Informatics for Biology, Bioindustry, Environment, Agriculture and Healthcare, University of Malaya, Kuala Lumpur, Malaysia
| | - Saad Tayyab
- Biochemistry Programme, Biomolecular Research Group, Institute of Biological Sciences, Faculty of Science, University of Malaya, Kuala Lumpur, Malaysia.,Centre of Research for Computational Sciences and Informatics for Biology, Bioindustry, Environment, Agriculture and Healthcare, University of Malaya, Kuala Lumpur, Malaysia
| |
Collapse
|
27
|
Nigam M, Suleria HAR, Farzaei MH, Mishra AP. Marine anticancer drugs and their relevant targets: a treasure from the ocean. Daru 2019; 27:491-515. [PMID: 31165439 PMCID: PMC6593002 DOI: 10.1007/s40199-019-00273-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 05/08/2019] [Indexed: 02/07/2023] Open
Abstract
Marine organisms comprising animals and plants are wealthiest sources of bioactive compounds possessing various pharmacological properties specifically: free radical scavenging, antitumor, antimicrobial, analgesic, neuroprotective and immunomodulatory. Marine drugs provide an alternative source to meet the demand of effective, safe and low-cost drugs that are rising with the continuously growing world population. Cancer is one of the leading reasons of mortality in western nations in contrast to communicable diseases of developing nations. In spite of outstanding developments in cancer therapy in past three decades, there is still an insistent necessity for innovative drugs in the area of cancer biology, especially in the unexplored area of marine anticancer compounds. However, recent technological innovations in structure revelation, synthetic creation of new compounds and biological assays have made possible the isolation and clinical assessment of innumerable unique anticancer compounds from marine environment. This review provides an insight into the anticancer research so far conducted in the area of the marine natural products/synthetic derivatives, their possible molecular targets and the current challenges in the drug development. Graphical abstract.
Collapse
Affiliation(s)
- Manisha Nigam
- Department of Biochemistry, H. N. B. Garhwal (A Central) University, Srinagar Garhwal, Uttarakhand 246174 India
| | - Hafiz Ansar Rasul Suleria
- Centre for Chemistry and Biotechnology, School of Life and Environmental Sciences, Deakin University, Pigdons Road, Waurn Ponds, Victoria 3216 Australia
- UQ Diamantina Institute, Translational Research Institute, Faculty of Medicine, The University of Queensland, 37 Kent Street Woolloongabba, Brisbane, QLD 4102 Australia
- Department of Food, Nutrition, Dietetics and Health, Kansas State University, Manhattan, KS 66506 USA
| | - Mohammad Hosein Farzaei
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Abhay Prakash Mishra
- Department of Pharmaceutical Chemistry, H. N. B. Garhwal (A Central) University, Srinagar Garhwal, Uttarakhand 246174 India
| |
Collapse
|
28
|
My journey from tyrosine phosphorylation inhibitors to targeted immune therapy as strategies to combat cancer. Proc Natl Acad Sci U S A 2019; 116:11579-11586. [PMID: 31076554 DOI: 10.1073/pnas.1816012116] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Since the 1980s there has been a drive toward personalized targeted therapy for cancer. "Targeted cancer therapy" originally focused on inhibiting essential tumor survival factors, primarily protein tyrosine kinases. The complexity and rapid mutability of tumors, however, enable them to develop resistance to tyrosine kinase inhibitors (TKIs), even when these are multitargeted or applied in combination. This has led to the development of targeted cancer immunotherapy, to enhance immune surveillance against the tumor. In this paper, we provide a personal view of the development of targeted therapy, from TKIs to targeted immunotherapy.
Collapse
|
29
|
Cardoso E, Csajka C, Schneider MP, Widmer N. Effect of Adherence on Pharmacokinetic/Pharmacodynamic Relationships of Oral Targeted Anticancer Drugs. Clin Pharmacokinet 2019. [PMID: 28634655 DOI: 10.1007/s40262-017-0571-z] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The emergence of oral targeted anticancer agents transformed several cancers into chronic conditions with a need for long-term oral treatment. Although cancer is a life-threatening condition, oncology medication adherence-the extent to which a patient follows the drug regimen that is intended by the prescriber-can be suboptimal in the long term, as in any other chronic disease. Poor adherence can impact negatively on clinical outcomes, notably because most of these drugs are given as a standard non-individualized dosage despite marked inter-individual variabilities that can lead to toxic or inefficacious drug concentrations. This has been especially studied with the prototypal drug imatinib. In the context of therapeutic drug monitoring (TDM), increasingly advocated for oral anticancer treatment optimization, unreported suboptimal adherence affecting drug intake history may lead to significant bias in the concentration interpretation and inappropriate dosage adjustments. In the same way, suboptimal adherence may also bias the results of pharmacokinetic modeling studies, which will affect in turn Bayesian TDM interpretation that relies on such population models. Detailed knowledge of the influence of adherence on plasma concentrations in pharmacokinetic studies or in routine TDM programs is however presently missing in the oncology field. Studies on this topic are therefore eagerly awaited to better pilot the treatment of cancer with the new targeted agents and to find their optimal dosage regimen. Hence, the development and assessment of effective medication adherence programs are warranted for these treatments.
Collapse
Affiliation(s)
- Evelina Cardoso
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Chantal Csajka
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland
| | - Marie P Schneider
- School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, Geneva, Switzerland.,Community Pharmacy, Department of Ambulatory Care and Community Medicine, University of Lausanne, Lausanne, Switzerland
| | - Nicolas Widmer
- Pharmacy of Eastern Vaud Hospitals, Vevey, Switzerland. .,Division of Clinical Pharmacology, Service of Biomedicine, Lausanne University Hospital, Rue du Bugnon 17, 1011, Lausanne, Switzerland.
| |
Collapse
|
30
|
Sheikhi M, Shahab S, Alnajjar R, Ahmadianarog M, Kaviani S. Investigation of Adsorption Tyrphostin AG528 Anticancer Drug Upon the CNT(6,6-6) Nanotube: A DFT Study. Curr Mol Med 2019; 19:91-104. [PMID: 30813875 DOI: 10.2174/1566524019666190226111823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/07/2018] [Accepted: 02/18/2019] [Indexed: 12/31/2022]
Abstract
OBJECTIVE In the present study, the interaction between drug Tyrphostin AG528 and CNT(6,6-6) nanotube by Density Functional Theory (DFT) calculations in solvent water has been investigated for the first time. METHODS AND RESULTS According to the calculations, intermolecular hydrogen bonds take place between an active position of the molecule Tyrphostin AG528 and hydrogen atoms of the nanotube which play an important role in the stability of complex CNT(6,6- 6)/Tyrphostin AG528. The non-bonded interaction effects of the molecule Tyrphostin AG528 with CNT(6,6-6) nanotube on the electronic properties, chemical shift tensors and natural charge have also been detected. The natural bond orbital (NBO) analysis suggested that the molecule Tyrphostin AG528 as an electron donor and the CNT(6,6-6) nanotube play the role of an electron acceptor at the complex CNT(6,6-6)/Tyrphostin AG528. CONCLUSION The electronic spectra of the Tyrphostin AG528 drug and complex CNT(6,6-6)/Tyrphostin AG528 in solvent water were calculated by Time-Dependent Density Functional Theory (TD-DFT) for the investigation of adsorption effect of the Tyrphostin AG528 drug over nanotube on maximum wavelength. Then, the possibility of the use of CNT(6,6-6) nanotube for Tyrphostin AG528 delivery to the diseased cells has been established.
Collapse
Affiliation(s)
- Masoome Sheikhi
- Young Researchers and Elite Club, Gorgan Branch, Islamic Azad University, Gorgan, Iran
| | - Siyamak Shahab
- Institute of Physical Organic Chemistry, National Academy of Sciences of Belarus,13 Surganov Str., Minsk 220072, Belarus.,Institute of Chemistry of New Materials, National Academy of Sciences of Belarus, 36 Skarina Str., Minsk 220141, Belarus.,Belarussian State University, ISEI BSU Minsk, Republic of Belarus
| | - Radwan Alnajjar
- Department of Chemistry, Faculty of Science, University of Benghazi, Benghazi, Libya.,Department of Chemistry, University of Cape Town, Rondebosch 7701, South Africa
| | - Mahin Ahmadianarog
- Department of Chemistry, Malekan Branch, Islamic Azad University, Malekan, Iran
| | - Sadegh Kaviani
- Department of Chemistry, Ferdowsi University of Mashhad, Mashhad, Iran
| |
Collapse
|
31
|
Sheikhhosseini E, Soltaninejad S. Design and Efficient Synthesis of Novel Biological Benzylidenemalononitrile Derivatives Containing Ethylene Ether Spacers. IRANIAN JOURNAL OF SCIENCE AND TECHNOLOGY, TRANSACTIONS A: SCIENCE 2019. [DOI: 10.1007/s40995-017-0376-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
32
|
Locke GM, Bernhard SSR, Senge MO. Nonconjugated Hydrocarbons as Rigid-Linear Motifs: Isosteres for Material Sciences and Bioorganic and Medicinal Chemistry. Chemistry 2019; 25:4590-4647. [PMID: 30387906 DOI: 10.1002/chem.201804225] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2018] [Revised: 10/20/2018] [Indexed: 01/02/2023]
Abstract
Nonconjugated hydrocarbons, like bicyclo[1.1.1]pentane, bicyclo[2.2.2]octane, triptycene, and cubane are a unique class of rigid linkers. Due to their similarity in size and shape they are useful mimics of classic benzene moieties in drugs, so-called bioisosteres. Moreover, they also fulfill an important role in material sciences as linear linkers, in order to arrange various functionalities in a defined spatial manner. In this Review article, recent developments and usages of these special, rectilinear systems are discussed. Furthermore, we focus on covalently linked, nonconjugated linear arrangements and discuss the physical and chemical properties and differences of individual linkers, as well as their application in material and medicinal sciences.
Collapse
Affiliation(s)
- Gemma M Locke
- School of Chemistry, SFI Tetrapyrrole Laboratory, Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, 152-160 Pearse Street, Dublin, 2, Ireland
| | - Stefan S R Bernhard
- School of Chemistry, SFI Tetrapyrrole Laboratory, Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, 152-160 Pearse Street, Dublin, 2, Ireland
| | - Mathias O Senge
- School of Chemistry, SFI Tetrapyrrole Laboratory, Trinity Biomedical Sciences Institute, Trinity College Dublin, The University of Dublin, 152-160 Pearse Street, Dublin, 2, Ireland
| |
Collapse
|
33
|
Riikonen R, Kokki H. CSF nerve growth factor (β-NGF) is increased but CSF insulin-like growth factor-(IGF-1) is normal in children with tuberous sclerosis and infantile spasms. Eur J Paediatr Neurol 2019; 23:191-196. [PMID: 30503720 DOI: 10.1016/j.ejpn.2018.11.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Revised: 11/01/2018] [Accepted: 11/01/2018] [Indexed: 01/31/2023]
Abstract
Tuberous sclerosis is associated with epilepsy that is often refractory. We examined cerebrospinal fluid (CSF) concentrations for neurotrophins, nerve growth factor (β-NGF) and insulin-like growth factor (IGF-1) in children with infantile spasms between 1997 and 2010. We classified the patients as follows: tuberous sclerosis (n = 5), cryptogenic spasms (n = 6), postinfectious spasms (n = 5) and other symptomatic spasms (n = 22). We had 22 age- and sex-matched controls for CSF-NGF and 14 for CSF-IGF-1. The median of CSF-NGF was higher in those with tuberous sclerosis, 56 (minimum-maximum, 8.0-131) ng/L, in relative to age- and sex-matched controls, 6.7 (0.0-22) ng/L, and symptomatic infantile spasms, 0.0 (0.0-4.5) ng/L or cryptogenic cases of infantile spasms, 6.2 (3.9-8.8) ng/L, respectively. CSF-NGF were highest in children with postinfectious aetiology, 408 (89-778) ng/L. CSF-IGF-1 of tuberous sclerosis, 0.65 (0.35-0.98) μg/L, did not differ from the cryptogenic spasms, 0.68 (0.32-0.87) μg/L, or from age- and sex-matched controls 0.52 (0.22-0.77) μg/L. Patients with tuberous sclerosis and cryptogenic spasms had normal development prior the ACTH therapy. We suggest that increased CSF-NGF might indicate a persistent activation of inflammatory pathways in cortical tubers in tuberous sclerosis and this would reflect in CSF concentrations.
Collapse
Affiliation(s)
- Raili Riikonen
- Children's Hospital, University of Eastern Finland and Kuopio University Hospital, Kuopio, Finland.
| | - Hannu Kokki
- School of Medicine, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
34
|
Türe A, Kahraman DC, Cetin-Atalay R, Helvacıoğlu S, Charehsaz M, Küçükgüzel İ. Synthesis, anticancer activity, toxicity evaluation and molecular docking studies of novel phenylaminopyrimidine-(thio)urea hybrids as potential kinase inhibitors. Comput Biol Chem 2018; 78:227-241. [PMID: 30579980 DOI: 10.1016/j.compbiolchem.2018.12.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2018] [Revised: 12/07/2018] [Accepted: 12/08/2018] [Indexed: 12/15/2022]
Abstract
Thirty-two novel urea/thiourea compounds as potential kinase inhibitor were designed, synthesized and evaluated for their cytotoxic activity on breast (MCF7), colon (HCT116) and liver (Huh7) cancer cell lines. Compounds 10, 19 and 30 possessing anticancer activity with IC50 values of 0.9, 0.8 and 1.6μM respectively on Huh7 cells were selected for further studies. These hit compounds were tested against liver carcinoma panel. Real time cell electronic sensing assay was used to evaluate the effects of the compounds 10, 19 and 30 on the growth pattern of liver cancer cells. Apoptotic cell death and cell cycle analysis upon treatment of liver carcinoma cells with hit compounds were determined. A significant apoptotic cell death was detected upon treatment of Huh7 and Mahlavu cells with compound 30 after 48 h of treatment. Additionally, compound 10 caused cell cycle arrest at G0/G1 phase. Mutagenicity of hit compounds was evaluated. Assertively, these compounds were not found to be mutagenic on Salmonella typhimurium strains TA98 and TA100. To understand the binding modes of the synthesized compounds, molecular docking studies were performed using the crystal data of VEGFR and Src-kinase enzymes in correlation with anticancer activities.
Collapse
Affiliation(s)
- Aslı Türe
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668 İstanbul, Turkey
| | - Deniz Cansen Kahraman
- Cancer System Biology Laboratory (CanSyL), Graduate School of Informatics, Middle East Technical University, 06800 Ankara, Turkey
| | - Rengul Cetin-Atalay
- Cancer System Biology Laboratory (CanSyL), Graduate School of Informatics, Middle East Technical University, 06800 Ankara, Turkey
| | - Sinem Helvacıoğlu
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Ataşehir, 34750 İstanbul, Turkey
| | - Mohammad Charehsaz
- Department of Toxicology, Faculty of Pharmacy, Yeditepe University, Ataşehir, 34750 İstanbul, Turkey
| | - İlkay Küçükgüzel
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Marmara University, Haydarpaşa, 34668 İstanbul, Turkey.
| |
Collapse
|
35
|
Yan LH, Chen ZN, Li CJ, Chen J, Qin YZ, Chen JS, Tang WZ. Prolonging Gastrointestinal-Stromal-Tumor-free life, an optimal suggestion of imatinib intervention ahead of operation. J Cancer 2018; 9:3850-3857. [PMID: 30410587 PMCID: PMC6218762 DOI: 10.7150/jca.25263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Accepted: 04/14/2018] [Indexed: 01/16/2023] Open
Abstract
Background: Imatinib has been regarded as the first successful synthetic small molecule targeting at blocking tyrosine kinase. Its high efficacy stabilized disease in above 80% of chronic myeloid leukemia (CML) patients over 10 years survival. Due to the similar canceration of gastrointestinal stromal tumor (GIST) as to CML, imatinib has been approved to be used as first-line treatment. Study design: Our retrospective study was proposed to enroll 191 GIST patients with larger tumor size (≥8 cm) who preoperative accepted imatinib from those with direct operation. Analysis included demographics, cancer specific survival and relationship of their risk factors. Results: Male patients and gastrointestinal (GI) tract location took higher proportion in total cases, detection of KIT mutant took 89.7% among all traceable genetic testing. Patients with preoperative imatinib can achieve higher cancer specific survival (CSS) after both in 1 year and 3 years duration than their counterpart. Tumor size above its threshold of 8 cm would be a hazardous factor for poor prognosis. Conclusion: In conclusion, as for regressing tumor progression and creating operative chance, preoperative imatinib should be considered for the patients with high risk, although the precise duration of this intervention needs further validation.
Collapse
Affiliation(s)
- Lin-Hai Yan
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Zhi-Ning Chen
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Chun-Jun Li
- Department of Pathology, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jia Chen
- Department of Medical Image Center, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Yu-Zhou Qin
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Jian-Si Chen
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| | - Wei-Zhong Tang
- Department of Gastrointestinal Surgery, Affiliated Tumor Hospital of Guangxi Medical University, Nanning 530021, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
36
|
Distinct phosphorylation sites/clusters in the carboxyl terminus regulate α 1D-adrenergic receptor subcellular localization and signaling. Cell Signal 2018; 53:374-389. [PMID: 30419287 DOI: 10.1016/j.cellsig.2018.11.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 11/08/2018] [Accepted: 11/08/2018] [Indexed: 12/28/2022]
Abstract
The human α1D-adrenergic receptor is a seven transmembrane-domain protein that mediates many of the physiological actions of adrenaline and noradrenaline and participates in the development of hypertension and benign prostatic hyperplasia. We recently reported that different phosphorylation patterns control α1D-adrenergic receptor desensitization. However, to our knowledge, there is no data regarding the role(s) of this receptor's specific phosphorylation residues in its subcellular localization and signaling. In order to address this issue, we mutated the identified phosphorylated residues located on the third intracellular loop and carboxyl tail. In this way, we experimentally confirmed α1D-AR phosphorylation sites and identified, in the carboxyl tail, two groups of residues in close proximity to each other, as well as two individual residues in the proximal (T442) and distal (S543) regions. Our results indicate that phosphorylation of the distal cluster (T507, S515, S516 and S518) favors α1D-AR localization at the plasma membrane, i. e., substitution of these residues for non-phosphorylatable amino acids results in the intracellular localization of the receptors, whereas phospho-mimetic substitution allows plasma membrane localization. Moreover, we found that T442 phosphorylation is necessary for agonist- and phorbol ester-induced receptor colocalization with β-arrestins. Additionally, we observed that substitution of intracellular loop 3 phosphorylation sites for non-phosphorylatable amino acids resulted in sustained ERK1/2 activation; additional mutations in the phosphorylated residues in the carboxyl tail did not alter this pattern. In contrast, mobilization of intracellular calcium and receptor internalization appear to be controlled by the phosphorylation of both third-intracellular-loop and carboxyl terminus-domain residues. In summary, our data indicate that a) both the phosphorylation sites present in the third intracellular loop and in the carboxyl terminus participate in triggering calcium signaling and in turning-off α1D-AR-induced ERK activation; b) phosphorylation of the distal cluster appears to play a role in receptor's plasma membrane localization; and c) T442 appears to play a critical role in receptor phosphorylation and receptor-β-arrestin colocalization.
Collapse
|
37
|
Aschner Y, Downey GP. The Importance of Tyrosine Phosphorylation Control of Cellular Signaling Pathways in Respiratory Disease: pY and pY Not. Am J Respir Cell Mol Biol 2018; 59:535-547. [PMID: 29812954 PMCID: PMC6236691 DOI: 10.1165/rcmb.2018-0049tr] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/23/2018] [Indexed: 01/02/2023] Open
Abstract
Reversible phosphorylation of proteins on tyrosine residues is an essential signaling mechanism by which diverse cellular processes are closely regulated. The tight temporal and spatial control of the tyrosine phosphorylation status of proteins by protein tyrosine kinases (PTKs) and protein tyrosine phosphatases (PTPs) is critical to cellular homeostasis as well as to adaptations to the external environment. Via regulation of cellular signaling cascades involving other protein kinases and phosphatases, receptors, adaptor proteins, and transcription factors, PTKs and PTPs closely control diverse cellular processes such as proliferation, differentiation, migration, inflammation, and maintenance of cellular barrier function. Given these key regulatory roles, it is not surprising that dysfunction of PTKs and PTPs is important in the pathogenesis of human disease, including many pulmonary diseases. The roles of various PTKs and PTPs in acute lung injury and repair, pulmonary fibrosis, pulmonary vascular disease, and inflammatory airway disease are discussed in this review. It is important to note that although there is overlap among many of these proteins in various disease states, the mechanisms by which they influence the pathogenesis of these conditions differ, suggesting wide-ranging roles for these enzymes and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Yael Aschner
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, and
| | - Gregory P. Downey
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, and
- Department of Immunology and Microbiology, University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, Colorado; and
- Department of Medicine
- Department of Pediatrics, and
- Department of Biomedical Research, National Jewish Health, Denver, Colorado
| |
Collapse
|
38
|
D'Andrea P, Sciancalepore M, Veltruska K, Lorenzon P, Bandiera A. Epidermal Growth Factor - based adhesion substrates elicit myoblast scattering, proliferation, differentiation and promote satellite cell myogenic activation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1866:504-517. [PMID: 30343052 DOI: 10.1016/j.bbamcr.2018.10.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 10/12/2018] [Accepted: 10/16/2018] [Indexed: 12/19/2022]
Abstract
The biochemical properties of muscle extracellular matrix are essential for stem cell adhesion, motility, proliferation and myogenic development. Recombinant elastin-like polypeptides are synthetic polypeptides that, besides maintaining some properties of the native protein, can be tailored by fusing bioactive sequences to their C-terminal. Our laboratory synthesized several Human Elastin-Like Polypeptides (HELP) derived from the sequence of human tropoelastin. Here, we developed a novel HELP family member by fusing the elastin-like backbone to the sequence of human Epidermal Growth Factor. We employed this synthetic protein, named HEGF, either alone or in combination with other proteins of the HELP family carrying RGD-integrin binding sites, as adhesion substrate for C2C12 myoblasts and satellite cells primary cultures. Adhesion of myoblasts to HEGF-based substrates induced scattering, decreased adhesion and cytoskeleton assembly; the concomitant presence of the RGD motifs potentiated all these effects. Recombinant substrates induced myoblasts proliferation, differentiation and the development of multinucleated myotubes, thus favoring myoblasts expansion and preserving their myogenic potential. The effects induced by adhesion substrates were inhibited by AG82 (Tyrphostin 25) and herbimycin A, indicating their dependence on the activation of both the EGF receptor and the tyrosine kinase c-src. Finally, HEGF increased the number of muscle stem cells (satellite cells) derived from isolated muscle fibers in culture, thus highlighting its potential as a novel substrate for skeletal muscle regeneration strategies.
Collapse
Affiliation(s)
- Paola D'Andrea
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy.
| | - Marina Sciancalepore
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy; Centre for Neuroscience B.R.A.I.N., University of Trieste, I-34127 Trieste, Italy
| | - Katerina Veltruska
- Department of Surface and Plasma Science, Faculty of Mathematics and Physics, Charles University in Prague V Holešovičkách 747/2, Praha 8, Czech Republic
| | - Paola Lorenzon
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy; Centre for Neuroscience B.R.A.I.N., University of Trieste, I-34127 Trieste, Italy
| | - Antonella Bandiera
- Department of Life Sciences, University of Trieste, I-34127 Trieste, Italy
| |
Collapse
|
39
|
Kim M, Baek M, Kim DJ. Protein Tyrosine Signaling and its Potential Therapeutic Implications in Carcinogenesis. Curr Pharm Des 2018. [PMID: 28625132 DOI: 10.2174/1381612823666170616082125] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Protein tyrosine phosphorylation is a crucial signaling mechanism that plays a role in epithelial carcinogenesis. Protein tyrosine kinases (PTKs) control various cellular processes including growth, differentiation, metabolism, and motility by activating major signaling pathways including STAT3, AKT, and MAPK. Genetic mutation of PTKs and/or prolonged activation of PTKs and their downstream pathways can lead to the development of epithelial cancer. Therefore, PTKs became an attractive target for cancer prevention. PTK inhibitors are continuously being developed, and they are currently used for the treatment of cancers that show a high expression of PTKs. Protein tyrosine phosphatases (PTPs), the homeostatic counterpart of PTKs, negatively regulate the rate and duration of phosphotyrosine signaling. PTPs initially were considered to be only housekeeping enzymes with low specificity. However, recent studies have demonstrated that PTPs can function as either tumor suppressors or tumor promoters, depending on their target substrates. Together, both PTK and PTP signal transduction pathways are potential therapeutic targets for cancer prevention and treatment.
Collapse
Affiliation(s)
- Mihwa Kim
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Minwoo Baek
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Dae Joon Kim
- Department of Biomedical Sciences, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, USA
| |
Collapse
|
40
|
Hiller NDJ, Silva NAAE, Faria RX, Souza ALA, Resende JALC, Borges Farias A, Correia Romeiro N, de Luna Martins D. Synthesis and Evaluation of the Anticancer and Trypanocidal Activities of Boronic Tyrphostins. ChemMedChem 2018; 13:1395-1404. [PMID: 29856519 DOI: 10.1002/cmdc.201800206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/14/2018] [Indexed: 12/13/2022]
Abstract
Molecules containing an (cyanovinyl)arene moiety are known as tyrphostins because of their ability to inhibit proteins from the tyrosine kinase family, an interesting target for the development of anticancer and trypanocidal drugs. In the present work, (E)-(cyanovinyl)benzeneboronic acids were synthesized by Knoevenagel condensations without the use of any catalysts in water through a simple protocol that completely avoided the use of organic solvents in the synthesis and workup process. The in vitro anticancer and trypanocidal activities of the synthesized boronic acids were also evaluated, and it was discovered that the introduction of the boronic acid functionality improved the activity of the boronic tyrphostins. In silico target fishing with the use of a chemogenomic approach suggested that tyrosine-phosphorylation-regulated kinase 1a (DYRK1A) was a potential target for some of the designed compounds.
Collapse
Affiliation(s)
- Noemi de J Hiller
- Research Group on Catalysis and Synthesis (CSI), Universidade Federal Fluminense, Laboratório 413, Instituto de Química, Campus do Valonguinho, Centro, Niterói, RJ, 24020-141, Brazil
| | - Nayane A A E Silva
- Research Group on Catalysis and Synthesis (CSI), Universidade Federal Fluminense, Laboratório 413, Instituto de Química, Campus do Valonguinho, Centro, Niterói, RJ, 24020-141, Brazil
| | - Robson X Faria
- Laboratory of Toxoplasmosis and other Protozoan Diseases, Oswaldo Cruz Institute (Fiocruz), Brasil
| | - André Luís A Souza
- Laboratory of Biochemistry of Peptides, Oswaldo Cruz Institute (Fiocruz), Brazil
| | - Jackson A L C Resende
- Laboratory of Solid-State Chemistry, Universidade Federal do Mato Grosso, Instituto de Ciências Exatas e da Terra, Campus Universitário do Araguaia, Barra do Garças, MT, 78600-000, Brazil
| | - André Borges Farias
- Núcleo de Pesquisas em Ecologia e Desenvolvimento Social (NUPEM), Universidade Federal do Rio de Janeiro, Campus de Macaé, Av. Rotary Club s/n; São José do Barreto, Macaé, RJ, 27901-000, Brazil
| | - Nelilma Correia Romeiro
- Núcleo de Pesquisas em Ecologia e Desenvolvimento Social (NUPEM), Universidade Federal do Rio de Janeiro, Campus de Macaé, Av. Rotary Club s/n; São José do Barreto, Macaé, RJ, 27901-000, Brazil
| | - Daniela de Luna Martins
- Research Group on Catalysis and Synthesis (CSI), Universidade Federal Fluminense, Laboratório 413, Instituto de Química, Campus do Valonguinho, Centro, Niterói, RJ, 24020-141, Brazil
| |
Collapse
|
41
|
Wang J, Han Y, Shi X, Li Q, Zhang P, Yuan P, Ma F, Luo Y, Cai R, Fan Y, Chen S, Li Q, Xu B. Phase I safety and pharmacokinetic study of cipatinib, an original dual tyrosine kinase inhibitor. Thorac Cancer 2018; 9:1041-1047. [PMID: 29893055 PMCID: PMC6068438 DOI: 10.1111/1759-7714.12784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Revised: 05/14/2018] [Accepted: 05/15/2018] [Indexed: 11/29/2022] Open
Abstract
Background Cipatinib is a novel tyrosine kinase inhibitor against both EGFR and HER2/neu. This phase I trial was conducted to assess the safety, dose‐limiting toxicities (DLTs), and maximum‐tolerated dose of cipatinib in HER2‐positive patients with advanced breast cancer. Methods Eligible adults with advanced breast cancer were administered cipatinib 200 mg/day (n = 3) as an initial dose, with escalating dosages of 400 mg (n = 4), 800 mg (n = 2), 1200 mg (n = 3), 1400 mg (n = 3), 1600 mg (n = 3), and 1800 mg (n = 2) in 21 day cycles. DLTs were monitored until the end of cycle 2. Physical examinations, vital signs, blood sampling for hematology, clinical chemistry, and pharmacokinetics were performed throughout the trial. Results Of the 26 subjects enrolled, 23 completed the trial. A total of 143 adverse events (AEs) were reported, of which 87 were associated with cipatinib treatment and comprised: neutropenia (38%), hypertriglyceridemia (15%), fatigue (15%), nausea (12%), fever (19%), and myocardial ischemia (19%). Six AEs were graded 3–4 (neutropenia, increases in aspartate aminotransferase, and total bilirubin, fatigue, dizziness and nodal tachycardia), but none of the AEs observed were considered to be DLTs. Conclusion This tolerability study revealed that despite a mild toxicity profile, cipatinib was well tolerated up to the anticipated maximum dosage of 1800 mg/m2. Further clinical trials are warranted.
Collapse
Affiliation(s)
- Jiayu Wang
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yiqun Han
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiuqing Shi
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Li
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pin Zhang
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Yuan
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Ma
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yang Luo
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ruigang Cai
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ying Fan
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shanshan Chen
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qiao Li
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
42
|
Tyrphostin AG17 inhibits adipocyte differentiation in vivo and in vitro. Lipids Health Dis 2018; 17:128. [PMID: 29843731 PMCID: PMC5975476 DOI: 10.1186/s12944-018-0784-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Accepted: 05/18/2018] [Indexed: 12/28/2022] Open
Abstract
Background Excessive subcutaneous adiposity in obesity is associated to positive white adipocyte tissue (WAT) differentiation (adipogenesis) and WAT expandability. Here, we hypothesized that supplementation with the insulin inhibitor and mitochondrial uncoupler, Tyrphostin (T-AG17), in vitro and in vivo inhibits adipogenesis and adipocyte hypertrophy. Methods We used a 3T3-L1 proadipocyte cell line to identify the potential effect of T-AG17 on adipocyte differentiation and fat accumulation in vitro. We evaluated the safety of T-AG17 and its effects on physiological and molecular metabolic parameters including hormonal profile, glucose levels, adipogenesis and adipocyte hypertrophy in a diet-induced obesity model using C57BL/6 mice. Results We found that T-AG17 is effective in preventing adipogenesis and lipid synthesis in the 3T3-L1 cell line, as evidenced by a significant decrease in oil red staining (p < 0.05). In obese C57BL/6 mice, oral administration of T-AG17 (0.175 mg/kg for 2 weeks) lead to decreased fat accumulation and WAT hypertrophy. Further, T-AG17 induced adipocyte apoptosis by activating caspase-3. In the hepatocytes of obese mice, T-AG17 promoted an increase in the size of lipid inclusions, which was accompanied by glycogen accumulation. T-AG17 did not alter serum biochemistry, including glucose, insulin, leptin, free fatty acids, creatinine, and aspartate aminotransferase. Conclusion T-AG17 promotes adipocyte apoptosis in vivo and is an effective modulator of adipocyte differentiation and WAT hypertrophy in vitro and in vivo. Therefore, T-AG17 may be useful as a pharmacological obesity treatment.
Collapse
|
43
|
Hassan W, Chitcholtan K, Sykes P, Garrill A. Ascitic fluid from advanced ovarian cancer patients compromises the activity of receptor tyrosine kinase inhibitors in 3D cell clusters of ovarian cancer cells. Cancer Lett 2018; 420:168-181. [PMID: 29432847 DOI: 10.1016/j.canlet.2018.02.013] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 01/31/2018] [Accepted: 02/06/2018] [Indexed: 12/18/2022]
Abstract
Ovarian cancer patients in the advanced stages of the disease show clinical ascites, which is associated with a poor prognosis. There is limited understanding of the effect of ascitic fluid on ovarian cancer cells and their response to anticancer drugs. We investigated the antitumour effects of EGFR/Her-2 (canertinib) and c-Met (PHA665752) inhibitors in a 3D cell model of three ovarian cancer lines. Single and combined inhibitor treatments affected cell growth of OVCAR-5 and SKOV-3 cell lines but not OV-90 cell line. Growth reduction was correlated with the down expression of PCNA, EGFR, HER-2, c-MET, ERK and AKT and their phosphorylation status in cells in growth factor supplemented media. However, these effects were not re-producible in OVCAR-5 and SKOV-3 cell lines when they were exposed to ascitic fluid obtained from three ovarian cancer patients. Serum albumin and protein components in the ascitic fluids may reduce the cellular uptake of the inhibitors.
Collapse
Affiliation(s)
- Wafaa Hassan
- School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, 8041, New Zealand.
| | - Kenny Chitcholtan
- Gynaecological Oncology Research Group, Department of Obstetrics and Gynaecology, University of Otago, Christchurch Women's Hospital, 2 Riccarton Avenue, Christchurch, 8011, New Zealand.
| | - Peter Sykes
- Gynaecological Oncology Research Group, Department of Obstetrics and Gynaecology, University of Otago, Christchurch Women's Hospital, 2 Riccarton Avenue, Christchurch, 8011, New Zealand.
| | - Ashley Garrill
- School of Biological Sciences, University of Canterbury, Private Bag 4800, Christchurch, 8041, New Zealand.
| |
Collapse
|
44
|
Thongon N, Castiglioni I, Zucal C, Latorre E, D'Agostino V, Bauer I, Pancher M, Ballestrero A, Feldmann G, Nencioni A, Provenzani A. The GSK3β inhibitor BIS I reverts YAP-dependent EMT signature in PDAC cell lines by decreasing SMADs expression level. Oncotarget 2018; 7:26551-66. [PMID: 27034169 PMCID: PMC5041998 DOI: 10.18632/oncotarget.8437] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 03/06/2016] [Indexed: 12/16/2022] Open
Abstract
The Yes-associated protein, YAP, is a transcriptional co-activator, mediating the Epithelial to Mesenchymal Transition program in pancreatic ductal adenocarcinoma (PDAC). With the aim to identify compounds that can specifically modulate YAP functionality in PDAC cell lines, we performed a small scale, drug-based screening experiment using YAP cell localization as the read-out. We identified erlotinib as an inducer of YAP cytoplasmic localization, an inhibitor of the TEA luciferase reporter system and the expression of the bona fide YAP target gene, Connective Tissue Growth Factor CTGF. On the other hand, BIS I, an inhibitor of PKCδ and GSK3β, caused YAP accumulation into the nucleus. Activation of β-catenin reporter and interfering experiments show that inhibition of the PKCδ/GSK3β pathway triggers YAP nuclear accumulation inducing YAP/TEAD transcriptional response. Inhibition of GSK3β by BIS I reduced the expression levels of SMADs protein and reduced YAP contribution to EMT. Notably, BIS I reduced proliferation, migration and clonogenicity of PDAC cells in vitro, phenocopying YAP genetic down-regulation. As shown by chromatin immunoprecipitation experiments and YAP over-expressing rescue experiments, BIS I reverted YAP-dependent EMT program by modulating the expression of the YAP target genes E-cadherin, vimentin, CTGF and of the newly identified target, CD133. In conclusion, we identified two different molecules, erlotinib and BIS I, modulating YAP functionality although via different mechanisms of action, with the second one specifically inhibiting the YAP-dependent EMT program in PDAC cell lines.
Collapse
Affiliation(s)
- Natthakan Thongon
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Ilaria Castiglioni
- Laboratory of Gene Expression and Muscular Dystrophy, San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Zucal
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Elisa Latorre
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Vito D'Agostino
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| | - Inga Bauer
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Michael Pancher
- High Throughput Screening Facility, Centre for Integrative Biology, University of Trento, Trento, Italy
| | | | - Georg Feldmann
- Laboratory of Pancreatic Cancer Translational Research, Clinic University of Bonn, Bonn, Germany
| | - Alessio Nencioni
- Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Alessandro Provenzani
- Laboratory of Genomic Screening, Centre for Integrative Biology, University of Trento, Trento, Italy
| |
Collapse
|
45
|
Synthesis and biological evaluation of new water-soluble photoactive chlorin conjugate for targeted delivery. Eur J Med Chem 2018; 144:740-750. [DOI: 10.1016/j.ejmech.2017.12.062] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 12/15/2017] [Accepted: 12/16/2017] [Indexed: 11/19/2022]
|
46
|
Athar M, Lone MY, Jha PC. Theoretical assessment of calix[ n ]arene as drug carriers for second generation tyrosine kinase inhibitors. J Mol Liq 2017. [DOI: 10.1016/j.molliq.2017.09.113] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
47
|
Childress ES, Alexopoulos SJ, Hoehn KL, Santos WL. Small Molecule Mitochondrial Uncouplers and Their Therapeutic Potential. J Med Chem 2017; 61:4641-4655. [DOI: 10.1021/acs.jmedchem.7b01182] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Elizabeth S. Childress
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Stephanie J. Alexopoulos
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
| | - Kyle L. Hoehn
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Kensington, NSW 2033, Australia
- Departments of Pharmacology and Medicine, Cardiovascular Research Center, and Emily Couric Clinical Cancer Center, University of Virginia, Charlottesville, Virginia 22908, United States
| | - Webster L. Santos
- Department of Chemistry and Virginia Tech Center for Drug Discovery, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
48
|
Elhenawy AA, Ashour RH, Nabih N, Shalaby NM, El-karef AA, Abou-El-Wafa HS. Insulin growth factor inhibitor as a potential new anti-schistosoma drug: An in vivo experimental study. Biomed Pharmacother 2017; 95:1346-1358. [DOI: 10.1016/j.biopha.2017.09.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2017] [Revised: 08/10/2017] [Accepted: 09/06/2017] [Indexed: 01/24/2023] Open
|
49
|
Lin B, Harris DR, Kirkman LMD, Perez AM, Qian Y, Schermerhorn JT, Hong MY, Winston DS, Xu L, Brandt GS. FIKK Kinase, a Ser/Thr Kinase Important to Malaria Parasites, Is Inhibited by Tyrosine Kinase Inhibitors. ACS OMEGA 2017; 2:6605-6612. [PMID: 30023525 PMCID: PMC6044879 DOI: 10.1021/acsomega.7b00997] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Accepted: 09/27/2017] [Indexed: 06/08/2023]
Abstract
A relatively high-affinity inhibitor of FIKK kinase from the malaria parasite Plasmodium vivax was identified by in vitro assay of recombinant kinase. The FIKK kinase family is unique to parasitic organisms of the Apicomplexan order and has been shown to be critical in malaria parasites. The recombinant kinase domain was expressed and screened against a small molecule library, revealing a number of tyrosine kinase inhibitors that block FIKK kinase activity. A family of tyrphostins was further investigated, to begin exploring the FIKK kinase pharmacophore. Finally, emodin was identified as a relatively high-affinity FIKK kinase inhibitor, identifying this family of anthraquinones as potential lead compounds for the development of antimalarials targeting the FIKK kinase.
Collapse
|
50
|
Lin BC, Harris DR, Kirkman LMD, Perez AM, Qian Y, Schermerhorn JT, Hong MY, Winston DS, Xu L, Lieber AM, Hamilton M, Brandt GS. The anthraquinone emodin inhibits the non-exported FIKK kinase from Plasmodium falciparum. Bioorg Chem 2017; 75:217-223. [PMID: 28987877 DOI: 10.1016/j.bioorg.2017.09.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/13/2017] [Accepted: 09/15/2017] [Indexed: 01/08/2023]
Abstract
The FIKK family of kinases is unique to parasites of the Apicomplexan order, which includes all malaria parasites. Plasmodium falciparum, the most virulent form of human malaria, has a family of 19 FIKK kinases, most of which are exported into the host red blood cell during malaria infection. Here, we confirm that FIKK 8 is a non-exported member of the FIKK kinase family. Through expression and purification of the recombinant kinase domain, we establish that emodin is a relatively high-affinity (IC50=2μM) inhibitor of PfFk8. Closely related anthraquinones do not inhibit PfFk8, suggesting that the particular substitution pattern of emodin is critical to the inhibitory pharmacophore. This first report of a P. falciparum FIKK kinase inhibitor lays the groundwork for developing specific inhibitors of the various members of the FIKK kinase family in order to probe their physiological function.
Collapse
Affiliation(s)
- Benjamin C Lin
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Darcy R Harris
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Lucy M D Kirkman
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Astrid M Perez
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Yiwen Qian
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Janse T Schermerhorn
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Min Y Hong
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Dennis S Winston
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Lingyin Xu
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Alexander M Lieber
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Matthew Hamilton
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States
| | - Gabriel S Brandt
- Department of Chemistry, Franklin & Marshall College, Lancaster, PA, United States.
| |
Collapse
|